You are on page 1of 16

International Journal of Nanomedicine Dovepress

open access to scientific and medical research

Open Access Full Text Article


REVIEW

The Influence of Nanoparticle Properties on Oral


Bioavailability of Drugs
This article was published in the following Dove Press journal:
International Journal of Nanomedicine downloaded from https://www.dovepress.com/ on 21-Oct-2023

International Journal of Nanomedicine

Yuanyuan Wang* Abstract: Oral administration has been the most common therapeutic regimen in various
Chao Pi* diseases because of its high safety, convenience, lower costs, and high compliance of patients.
Xianhu Feng However, susceptible in hostile gastrointestinal (GI) environment, many drugs show poor
Yi Hou permeability across GI tract mucus and intestinal epithelium with poor oral absorption and
limited therapeutic efficacy. In recent years, nanoparticulate drug delivery systems (NDDS) have
Ling Zhao
become a hot research spot because of their unique advantages including protecting drug from
Yumeng Wei
For personal use only.

premature degrading and interacting with the physiological environment, increasing intracellular
Central Nervous System Drug Key penetration, and enhancing drug absorption. However, a slight change in physicochemistry of
Laboratory of Sichuan Province, School
of Pharmacy, Southwest Medical nanoparticles can significantly impact their interaction with biological pathways and alter the
University, Luzhou, Sichuan 646000, oral bioavailability of drugs. Hence, this review focuses on the factors affecting oral bioavail­
People’s Republic of China ability from two aspects. On the one hand, the factors are the biochemical and physiological
*These authors contributed equally to barriers in oral drugs delivery. On the other hand, the factors are the nanoparticle properties
this work including size, surface properties, and shape of nanoparticles.
Keywords: oral bioavailability, nanoparticle properties, size, shape, surface properties

Introduction
Oral administration has been the most common manner of drug delivery for thousands
of years, with some advantages including the safety, it being well-tolerated, with low
treatment costs, good compliance, and convenience.1,2 Despite its outstanding advan­
tages, the traditionally oral administered drugs are faced with the daunting challenges
of poor and highly variable bioavailability, which can be frequently caused by inherent
instability and low solubility in variable conditions of the gastrointestinal (GI) tract,
poor permeability through GI barriers, and, in some cases, extensive pre-systemic
metabolism in the GI tract and liver (eg, cytochrome P-450).2,5
To surmount these barriers in the GI tract and significantly improve oral bioavail­
ability, designing and developing some effective oral drug delivery systems is quite
urgent.6 Thus, a large variety of new techniques and dosages of drugs have been
developed such as protein or polymer conjugates,7 solid drug dispersions,8 nanoparticle
technologies,9 and macroscopic systems such as capsules, gels, and films.10 Among
these strategies, nanoparticulate drug delivery systems (NDDS) have been attracting
more attention since they can protect the drug from premature degradation and inter­
action with the physiological environment, increase intracellular penetration, and
Correspondence: Ling Zhao; Yumeng Wei
Tel/Fax +86 830 3162292; enhance drug absorption (Figure 1).11,14 By far, numerous NDDS including albumin
+86 830 3162291 nanoparticles,15 chitosan (CS) nanoparticles,16 liposomes,17 polymeric micelles,18 etc.
Email zhaoling-998@163.com;
weiyumeng-268@163.com have been developed for the oral delivery of different drugs.

submit your manuscript | www.dovepress.com International Journal of Nanomedicine 2020:15 6295–6310 6295
DovePress © 2020 Wang et al. This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.
http://doi.org/10.2147/IJN.S257269
php and incorporate the Creative Commons Attribution – Non Commercial (unported, v3.0) License (http://creativecommons.org/licenses/by-nc/3.0/). By accessing the
work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For
permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms (https://www.dovepress.com/terms.php).
Wang et al Dovepress

Figure 1 The benefits of using nanoparticles.


Abbreviation: GI, gastrointestinal.

However, previous studies have reported that a slight variation in PH conditions, surfactants, and rich enzyme
change in physicochemistry of nanoparticles can significantly content in the GI tract, loose and firm mucus, and trans­
impact on their interaction with biological pathways and alter porters in vivo. Thus, it is vital to give an insight into the
the oral bioavailability of drugs.6,11,19 For example, the control obstacles for the development of oral drugs and the boost­
of dimension under 200 nm was more absorbed by the GI ing of oral bioavailability of drugs.27
tract.4,20,21 And other reports found that polystyrene (PS)
nanoparticles decorated with vitamin B12 (VitB12) were more
Biochemical Barriers
easily avoiding lysosomal digestion as well as entering epithe­
After oral administration, drugs would confront a harsh bio­
lial cells, compared with both soluble VitB12 ligand and
chemical environment,28 which comprises of PH variation in
unmodified nanoparticles.22 In addition to the influence of
the GI tract, metabolizing enzymes, surfactants like bile salt,
size and surface properties of nanoparticles, another paper
and the liver itself.4 The drugs can be destroyed by acidic
reported that oral bioavailability of drugs was also signifi­
environment, especially materials that are sensitive to acidic
cantly impacted by nanoparticle geometry,23–25 such as the
conditions.28 Furthermore, the variation in PH conditions is
fact that rod-nanoparticles had better oral bioavailability com­
from acidic to alkaline along the GI tract, which imposes
pared with sphere-nanoparticles.26 Therefore, it is necessary to
a hurdle to the stability of drugs and sometimes their
summarize the role of the size, surface properties, and shape of
carriers.29 In addition, surfactants in the GI tract also have
nanoparticle in oral drug delivery. We believe that understand­
a substantial impact on the structural integrity of drugs.29
ing the role of nanoparticle properties in GI tract transport as
Beyond these, the drugs after oral administration still have to
well as cellular uptake can bring forth a paradigm shift in
face a particularly rich enzyme content, composed of pro­
nanoparticle engineering for oral delivery. Hence, this review
teases, lipases, or amylases.30 These enzymes exist in the
focuses on the factors affecting oral bioavailability, in which
digestive tract, especially in the lower one, which are respon­
these factors are divided into two categories: the biochemical
sible for the metabolism.4 After entering intestinal epithelial
and physiological barriers in oral drugs delivery; and the
cells, the drugs will be threatened by extra-hepatic micro­
nanoparticle properties including size, surface properties, and
somal enzymes on the endoplasmic reticulum inside the
shape of nanoparticles (Figure 2).
cytoplasm liver (eg, the cytochrome P450 3A family of
phaseⅠ metabolic enzymes).31 In conclusion, the bioavail­
Physiological and Biochemical ability of drugs can be limited by the biochemical barriers
Disorders in Oral Delivery after oral administration. Hence, the numerous drugs are
Although oral drug delivery has been widely used in encapsulated to nanoparticles. Furthermore, the materials
clinical practice, oral bioavailability has always been lim­ used as the nanocarrier should be carefully chosen or chemi­
ited by the disorders of oral administration, including the cally modified adequately.28

6296 submit your manuscript | www.dovepress.com International Journal of Nanomedicine 2020:15


DovePress

Powered by TCPDF (www.tcpdf.org)


Dovepress Wang et al

Figure 2 (A) The nanoparticle properties including size, surface properties, and shape of nanoparticles. (B) Schematic representation illustrating the biochemical and
physiological barriers of oral drug delivery. (C) The relationship of the nanoparticle properties, oral bioavailability, and barriers in oral delivery.

Physiological Barriers interactions with the glycoproteins from mucus, limiting the
Mucus Barriers drugs absorption rate and decreasing the oral bioavailability
Some reports mentioned that drugs must be capable of pene­ of drugs.34 Meanwhile, hydrophobic bare globular regions
trating and crossing mucus barriers in order to be absorbed (non-glycosylated regions) with high density located on
and gain access to the circulatory system and target mucin chains generate multiple low-affinity adhesion inter­
tissue.32,33 The mucus layer is ubiquitous in the GI tract actions with hydrophobic regions of foreign particles.42 The
composed of water, proteins, lipids, electrolytes, antimicro­ large number of hydrophobic drugs through the mucus can be
bial peptides, sloughed epithelial cells, bile salts, and other rapidly removed, impairing drug effective therapy.42 In addi­
components available in the GI tract.34,35 Mucus can be tion, mucin molecules can form a network via disulfide
divided into two types: loose and firm mucus (Figure bonds. The non-glycosylated regions of the mucin molecule
3B).36 Loose mucus is easy to be removed by suction as are the site of interchain disulfide bridge that connects the
well as shear and high thickness of the layer.37,38 However, glycoprotein subunits.43 The mesh space and the brush-like
firm mucus adheres firmly or anchors to the epithelium sur­ structure of mucus can act as a size exclusion filter, reducing
face and is resistant to the removal by suction and shear.37 the mobility of large molecules, thereby increasing their
The main structural constituent of the mucus layer is mucin clearance rate.40,44,46 Another intriguing finding is that the
(Figure 3A) that can be defined as glycoproteins containing thickness of the mucosal barrier varies with the location of
heavily O-glycosylated serine/threonine-rich tandem repeat the GI region (Table 1).35,36 Crossing the mucus layer implies
domains.33,39 The oligosaccharide side chains including a particle size below 200 nm.28 Indeed, nanocarriers should
a terminal carboxyl group or ester sulfate groups give be small enough to avoid being blocked by the mucin mesh.
mucus its negative charge.33,40,41 Some positively charged Moreover, interactions between the surface of nanocarriers
drugs and their carriers have been shown to have electrostatic and mucus must be kept at a minimum.

submit your manuscript | www.dovepress.com


International Journal of Nanomedicine 2020:15 6297
DovePress

Powered by TCPDF (www.tcpdf.org)


Wang et al Dovepress

Figure 3 (A) Highly O-glycosylated mucin domains. Red, protein core; Green, oligosaccharides. (B) Schematic representation illustrating two types of mucus.
Abbreviations: Ser, serine; Thr, threonine; Pro, proline; Cys, cysteine; S-S, disulfide bonds; Ser-O-GalNAC, O-glycosylated serine.

Absorption Barriers of the Intestinal Epithelium patches.47,50 The transcellular pathway mainly absorbs par­
Intestinal epithelium with dense and orderly brush border on ticles through the cell membrane of enterocytes relying on
the top surface is an intrinsic physical barrier that consists of their size and hydrophobicity.50 In the absence of aggrega­
a single layer of columnar epithelial cells comprised of tion, the particles of smaller size are more easily absorbed by
absorptive (enterocytes) and secretory cells (goblet cells intestinal epithelial cells. It has been reported that a particle
and paneth cells).47,49 Epithelium permeation of drugs can size smaller than 300 nm could be untaken by enterocytes,
occur via transcellular route, paracellular avenues, and the while a size larger than 500 nm was more likely to be
densely clustered M cells located on the surface of Payer’s absorbed in jejuna Payer’s patches.50,52 The paracellular
pathway is another limiting step in transepithelial transport,
mediated through tight junctions (TJs), which is the way for
Table 1 Thickness of Mucus in Different Parts of the Human
the hydrophilic molecules uptake.49,50,53 Moreover, most
Gastrointestinal Tract
macromolecular drugs do not readily enter systemic circula­
Mucus Membrane Total Mucus Thickness (μm) Ref
tion through intercellular connections. It is reported that Ca2+
Gastric 30–300 [37] chelating agents could increase the paracellular permeability
Small Intestine 150–400 [37] by reversible opening of the tight junctions.54,55 Payer’s
Ileum 400–500 [36]
patches are actually the submucosal mass lymph node of
Colon 30–700 [36,37]
the small intestine, which are the main site of intestinal

6298 submit your manuscript | www.dovepress.com International Journal of Nanomedicine 2020:15


DovePress

Powered by TCPDF (www.tcpdf.org)


Dovepress Wang et al

mucosal immune induction. M cells located on the surface of cell monolayer (P<0.01) compared with NLC-200 nm and
Payer’s patches act as an entrance for drugs to the lymphatic NLC-300 nm.62 Moreover, pharmacokinetic studies shown
vessels, especially lipid soluble drugs, from the intestinal that NLC-100 nm exhibited highest Cmax and AUC com­
cavity to lymphoid cells, which shall reduce the primary pared with the others.62 Zhao et al63 also mentioned that
clearance of the liver.50 However, the lymphatic pathway the 100 nm nanovaccine exhibited better pharmacokinetic
shows only a high affinity for lipophilic compounds. efficacy than the 500 nm nanovaccine in the presence of
Ling et al56 increased the bioavailability of cefotaxime via alum adjuvant. Li et al62 concluded from their data that the
liposome preparation compared with aqueous solution and size of 100 nm might be the most suitable size for oral
the physical mixture. delivery of nanoparticles. However, the data of Banerjee
et al6 indicated that particle uptake by Caco-2 and Caco-2/
Transporters in vivo HT-29 cells was inversely related to their size, with uptake
In addition, there are a variety of transporters in vivo, such as of 50 nm > 200 nm > 500 nm > 1000 nm.6 Thus, the
P-glycoprotein (P-gp), cytoplasmic transporter, breast cancer hypothesis can be mentioned that nanoparticle size has
drug resistant proteins, and multidrug resistance associated a negative correlation influence on oral bioavailability of
protein, which release drugs from the intracellular environ­ drugs during the certain range. However, the optimal par­
ment to outside of the cell.4 P-gp encoded by multidrug ticle size still needs to be actively explored by researchers
resistance-1 gene is the major drug efflux transporter in oral drug delivery.
system.4 A lot of drugs (eg, paclitaxel (PTX), docetaxel,
etoposide, vinblastine, vincristine, and doxorubicin) are
known as potential substrates of efflux transporters. Various
The Effects of Surface Properties for
efflux transporters in vivo infinitely restrict oral absorption of Oral Bioavailability
drugs.4 Therefore, active modification of drug carriers is NDDS have many desirable characteristics for drug admin­
essential. istration such as improving solubility and stability of
drugs.64 However, even the most potent extended release
The Effect of Size for Oral nanoparticle would be rapidly cleared from the body if it
adheres to the superficial layers of mucus, reducing delivery
Bioavailability of the drug to the target tissues.65 The report demonstrated
It has been reported that the bioadhesion of solid lipid
that the surface chemistry of nanoparticles can greatly
nanoparticles (SLNs) in the GI tract can be improved via
impact their interaction with biological pathways and alter
smaller size to prolong their residence time and thus play
efficacy (Table 2).11 Efficient oral delivery of drugs based
a contributing role in improving oral bioavailability.4,57,58
on NDDS requires simultaneously overcoming physiologi­
Retinoic acid-SLNs showed an increase of 3-fold oral
cal and biochemical disorders of the body, which needs very
bioavailability of retinoic acid when particle size decreases
different or even contradictory surface properties of
from 328.8 nm to 89.3 nm.59 The fact proved that particle
nanocarriers.66 In general, it is a necessity to achieve
size plays a crucial role in oral drug delivery.60
a variety of treatment goals with different materials or
It has been concluded previously that particle size has
different modifications to nanoparticles. Thus, definitely
a great impact on the movement of particles in the mucus.
learning the role of surface properties of nanoparticles
For instance, Maisel et al61 found that particles of 40 and
about oral delivery is important.
100 nm in size spread over the mucosa reaching to the
deep, folded surfaces of the colorectal epithelium, whereas
particles 200 and 500 nm in size just spread over the The Particle Containing a Hydrophilic
surface. The data indicate that particles with smaller size Group
are easily reaching to the firm mucus that is resistant to the As previously discussed, after entering the GI tract
removal by suction and shear, thereby prolonging their through the esophagus, the oral drugs can be removed by
residence time. In addition, the cellular uptake and uptake non-covalent bonds (such as hydrophobic bonds, hydrogen
pathway of particles are also affected by the size of nano­ bonds, and electrostatic forces) with GI mucus, especially
particles. The smaller nanostructured lipid carriers (NLC- the superficial GI mucus, to prevent it entering the sys­
100 nm) show higher uptake efficiency in the Caco-2 cell temic circulation, thereby reducing the effective dosage of
(P<0.05) as well as higher permeation ability in Caco-2 drugs. Neutral hydrophilic materials (Figure 4) get more

submit your manuscript | www.dovepress.com


International Journal of Nanomedicine 2020:15 6299
DovePress

Powered by TCPDF (www.tcpdf.org)


Wang et al Dovepress

Table 2 The Effect of Various Surface Modification Materials of doxorubicin (DOX) loaded PLGA-Nanoparticles
Nanoparticles on the Oral Administration (PEGylated-DOX-PLGA-NPs) observed a 6.8-fold oral
Material Influential Effect Ref bioavailability as compared to DOX-S in pharmacokinetics
studies.68 In addition, Feeney et al69 modified lipid nanopar­
PEG Increasing mucus penetration (~5 KDa) [62]
ticles (LN) by using carrier materials of hydrogenated castor
Reduce recognition and clearance [37,71]
oil (HCO) and castor oil (CO) (containing PEG groups).
Enhance plasma circulation times [71] They found that the relative bioavailability (defined as the
Promote drug accumulation [37]
ratio of AUC 0–∞ for the modified LN and its structurally
analogous LN) of the formulations was 120% and 182% for
TA Improve solubility of hydrophobic [75]
HCO and CO formulations, respectively. However, some
molecules
articles also mentioned PEG had a strong viscosity, which
Inhibitory effect of P-gp [81] speculated hydrogen bonding between ether oxygen atoms in
Avoid gastric acid degradation [2] PEG and sugars on glycosylated mucins. Wang et al70
explored the physicochemical properties of PEG-coated
Polycation Increase solubility of hydrophobic [84]
molecules
nanoparticles, specifically PEG molecular weight and degree
of surface coverage, to reconcile the paradoxical reports of
PECs Facilitating mucus transport [43,85]
PEGs interactions with mucus. They explored the physico­
Increasing cellular penetration [89] chemical properties of PEG-coated nanoparticles which were
Lipid material Increase the lymphatic absorption [84]
formulated from PS modified PEGs of various molecular
(log P>5) Prevent the liver from first pass weights (2, 5, 10 kDa) to reconcile the paradoxical reports
metabolizing of PEGs interactions with mucus. They found that PS-
[84]
PEG2K(High) nanoparticles penetrated mucus with effective
speed only 7-fold reduced compared with in water. And they
Specific Increasing cellular uptake [96]
concluded that low molecular weight and high (dense) PEG
ligands
surface coverage was required for rapid mucus penetration of
Abbreviations: PEG, ploy(ethylene glycol); TA, tannic acid; PECs; polyelectrolyte
complexes; P-gp, P-glycoprotein.
coated particles, and that dense PEG coatings transferred
from being mucoinert to mucoadhesive when a critical mole­
cular weight existed between 5 and 10 kDa.
and more attention because of their difficulty in hydropho­
bic and electrostatic interaction with GI mucus. Such sub­
Coating Particles with Tannic Acid (TA)
stances that are easy to penetrate the mucus barrier are TA is a type of the natural polyphenols which is constituted
called mucopenetrating particles (MPP), which can plentiful catechol and pyrogallol moieties and generally recog­
improve epithelial distribution and retention time by pene­ nized as safe by the United States Food and Drug
trating the outer, rapidly clear mucus layers to reach the Administration (FDA).71,72 In recent years, it has been reported
more slowly cleared layers and increase the effective that TA can improve solubility of hydrophobic molecules by
amount of drugs into the systemic circulation.67 hydrogen bonding.73 Shen et al72 proved that TA anchored
onto the surface of hydrophobic PTX nanocore via multiple
PEGylated Drug Delivery Vehicles hydrogen bonding between pyrogallol or catechol groups of
The poly(ethylene glycol) (PEG), a passive mucopenetrating TA and hydroxyl groups or oxygen atoms of PTX molecules.
system, has been widely used in surface modification of The X-ray diffraction (XRD) pattern of the pure PTX powder
nanoparticles due to the property of reducing interactions showed a number of sharp Bragg peaks, corresponding to
with both luminal components and mucus in the gut.36 highly crystalline PTX.74 No discernible peaks appeared for
Maisel et al61 used PEG-modified poly(lactic-co-glycolic the TA-PTX complex, indicating the amorphous nature of the
acid) (PLGA) nanoparticles to study the mucus-penetrating complex coating.72 The data of release kinetics indicated that
of particles under different conditions (eg, providing normal TA-PTX complex nanoparticles displayed a gradual release
and colitis mice model, or increasing the volume of gavage), profile without burst release at an initial stage and finally
and concluded that the particles were uniformly distributed reached 90% of drug release after 36 hours.72 The good dis­
on the mucosa of epithelial cells. In the PEGylated solution behavior can be attributed to their amorphous nature

6300 submit your manuscript | www.dovepress.com International Journal of Nanomedicine 2020:15


DovePress

Powered by TCPDF (www.tcpdf.org)


Dovepress Wang et al

Figure 4 Structural formula of partial materials containing hydrophilic groups.

of PTX in the complex nanoparticles, which was helpful to to 85% optical transmittance after adjusting to pH 7.4. The
improve bioavailability.75 In addition, TA has been verified to particles’ diameter immediately increased from 54 nm to about
exhibit high biological activities,76,77 such as inhibited effect of 2 µm after dispersing in pH 2, while reduced to 70 nm in pH
P-gp. It is all known that P-gp-mediated efflux is one of the 7.4. In addition, it was worth mentioning that microaggregates
major absorptive barriers in vivo.78 Kitagawa et al79 demon­ can be found in pH 2, and the approximate recovery of particle
strated that TA displayed the P-gp inhibitory function by size to original state of TA-PTX nanoparticles when pH was
ATPase inhibition. Some scholars believed that nanoparticles adjusted to 6.8 or 7.4. These results suggested that TA-PTX
conjugated TA can avoid gastric acid degradation, achieve PH nanoparticles can reduce the burst release of drugs, possibly
dependent intestinal site release, and increase oral absorption prevent the acidic degradation of PTX in the stomach, and
of drugs.2 They rationally developed a hydrogen-bonded TA achieve intestinal site-specific drug release through physiolo­
acid-based nanoparticle, TA-PTX nanoparticles, via a flash gical pH stimulation.
nanoprecipitation process, and explored PH-sensitive beha­
viors of TA-PTX nanoparticles at different pH conditions. Coating Particles with Other Hydrophilic Substance
They found that the TA-PTX nanoparticles solution exhibited A great number of other hydrophilic substances are
strong opaque property at acidic conditions and then recovered abounding in nature, such as polyethylene oxide (PEO),

submit your manuscript | www.dovepress.com


International Journal of Nanomedicine 2020:15 6301
DovePress

Powered by TCPDF (www.tcpdf.org)


Wang et al Dovepress

poloxamer 188, and pluronic F-127 (PF-127). To enhance gelation method between the polycationic CS and sodium
mucus permeation and promote cellular uptake compared tripolyphosphate pentabasic anions, which were a potential
with cabazitaxel (CTX), Ren et al80 formulated CTX poly­ delivery system for the oral administration compared with
mer–lipid hybrid nanoparticles (CTX-PMONP). The nano­ IFNα.85 Dyawanapelly et al86 developed PLGA nanopar­
carriers were comprised of a poly(ε-caprolactone) (PCL) ticles, surface-modified chitosan oligosaccharide (COS)-
and chain triglyceride hybrid core for drug loading, and PLGA nanoparticles, and CS-PLGA nanoparticles and
encased in PEO shell by insertion of poloxamer 188.80 The compared mucoadhesion of CS and COS surface-
oral bioavailability of CTX was elevated from 7.7% CTX modified polymer nanoparticles for mucosal delivery of
solution (CTX-Sol) to 56.6% after oral administration of proteins. Their data indicated that positively charged COS-
CTX-PMONPs, approximately 7.3-times higher than that PLGA nanoparticles and CS-PLGA nanoparticles exhib­
of CTX-Sol.80 In addition, Li et al81 found that when the ited higher mucoadhesion than negatively charged PLGA
mucoinert polymer PF-127 was incorporated, the mucus nanoparticles. Wang et al87 also prepared PLGA nanopar­
penetrating properties of liposomes were improved signif­ ticle coating with CS and performed cellular uptake
icantly. PF127-inlaid liposomes and PF127-adsorbed lipo­ mechanisms as well as transmembrane permeability in
somes were prepared by a thin-film hydration method Madin-Darby canine kidney-cell monolayers. They found
followed by extrusion, in which coumarin 6 was loaded
that, under all conditions, CS-PLGA nanoparticles showed
as a fluorescence marker.81 A modified Franz diffusion cell
a greater potential to be transported into cells.
mounted with the intestinal mucus of rats was used to
Furthermore, pharmacokinetic studies demonstrated
study the diffusion characteristics of the two types of
marked improvement of 3.53-fold and 8.03-fold in
PF127 liposomes.81 The diffusion efficiency of the two
Wistar rat’s plasma as well as brain higher oral bioavail­
types of PF127-modified liposomes through intestinal rat
ability through CS-coated-Irinotecan (IRN)-loaded-PLGA
mucus was 5-7-fold higher than that of unmodified lipo­
nanoparticles when compared with IRN solution.88
somes. In general, hydrophilic materials are easy to pene­
trate the mucus barrier because of their difficulty in
hydrophobic and electrostatic interaction with GI mucus, The Particle Containing Lipid Material
thereby increasing the effective amount of drugs into the Group
systemic circulation. The majority of orally administered drugs are absorbed
into the portal blood to reach the systemic circulation.
The Particle Containing Polycationic However, the lipophilic substances (log P>5) usually
reach the systemic circulation through the lymphatic
Group
pathway.60 In addition, some papers have mentioned that
Formulation drugs with polycationic and surface-modified
lipophilic substances and macromolecular substances with
polycation particles have been shown to increase solubi­
lity, protect labile compounds from pH changes, and diges­ large molecular weights can increase the lymphatic
tive enzymes.82 And some studies also illustrated that absorption of drugs.89 Moreover, the permeability of lym­
apart from uncharged nanoparticles with hydrophilic and phatic vessels to nanoparticles is significantly higher than
a low hydrogen bonding capability, a densely charged that of capillaries.90 In addition, absorption of drugs
nanoparticle yet net neutral surface can be also considered through lymphatic vessels, due to its unique physiological
as MPP. These particles with neutral surface, high surface and anatomical structure, can prevent the first pass meta­
charge density of polyelectrolyte complexes (PECs), can bolizing of liver.60,91 Therefore, through lymphatic absorp­
create a hydrophilic surface that decreases its hydrophobic tion, the bioavailability of oral drugs can be significantly
interactions with mucus and facilitate mucus transport.42,83 increased.92 However, different lengths of fatty acid chains
CS, an attractive polymer, has been extensively used in have different absorption effects. The fatty acid chains
oral delivery. The combination of CS and chondroitin from c-14 to c-18 promote greater lymphatic
sulfate was used to prepare PECs that could penetrate absorption.93 Khoo et al94 also found that long-chain tri­
mucus to the higher extent compared with reference glycerides promoted absorption more effectively than
nanoparticles.84 In addition, interferon alpha (IFNα)- medium-chain triglycerides. Moreover, certain classes of
loaded CS-nanoparticles were prepared by the ionotropic phospholipids and surfactants used in the formulations of

6302 submit your manuscript | www.dovepress.com International Journal of Nanomedicine 2020:15


DovePress

Powered by TCPDF (www.tcpdf.org)


Dovepress Wang et al

liposomes can suppress the P-gp efflux system, thereby The Effect of Shape for Oral
increased absorption of drugs.4 Bioavailability
In addition to surface properties and size of nanoparticles,
The Particle Containing Specific Ligands the shape of nanoparticles has a great influence on their
Nanoparticle coated specific ligand has better intracellular interaction with biological systems, including cellular
uptake trend. For instance, in order to study the effect of uptake, plasma circulation and organ distribution and so
folate functionalization on the extracellular transport of PTX, on. However, understanding of how shape of nanoparticles
a chemotherapy drug with poor oral bioavailability, Roger affects biological systems is far from complete. Nature
et al95 modified PTX-PLGA nanoparticles with folate. Using abounds in viruses and bacteria of various forms.99 It is
the Caco-2 monolayer as an in vitro model, confocal micro­ well known that bacterial forms have an evolutionary
scopy showed that PTX loaded in PLGA nanoparticles advantage due to their interactions with surface mechan­
increased the apparent permeability in Caco-2 cells by isms such as passive diffusion and active transport.100 In
Darwinian evolution, changes in bacterial or viral form
5-times compared with free PTX. Functionalization of nano­
precede in function due to random mutations. There is
particles with folic acid further increased the transport capa­
a strong case for natural selection of bacteria and viruses
city (8-fold compared with free PTX).
with specific forms. In the future of medicine, nanoparti­
As we all know, the pharmacokinetic process of cargos
cles could also be shaped to suit their function.101
in vivo is very complicated, and it is difficult to achieve
Most current NDDS used in laboratory-scale or clinical
effective drug delivery via modifying nanoparticles with
trials are spherical due to ease of synthesis. However, nowa­
only one kind of material. Therefore, nowadays, many studies days, compounds in large numbers are being presented in
modify or prepare nanoparticles with different or even oppo­ different forms of nanoparticles. For instance, Wang et al102
site materials. Netsomboon et al36 mentioned that negatively synthesized gold nanoparticles through one pot synthesis
charged particles being capable of changing zeta-potential to with inorganic metal material Au. Different morphologies
a positive value once having permeated the mucus and having could be obtained by changing the gold to calcium ion
reached the epithelium might be a promising strategy. concentration ratios during the synthesis. As the calcium
Negatively charged and uncharged particles can easily move concentration decreased, the morphology changed from
through the mucus,32,96 whereas positively charged particles hexagons to pentagons and to triangles. Similarly, Huang
show a comparatively much higher cell uptake via endocyto­ et al19 fabricated different shaped MSNs (spheres, short rods
sis than negatively charged particles.97 A typical example was and long rods), via inorganic nonmetallic material, to study
that, after incubation with intestinal alkaline phosphatase, zeta the effect of MSNs on cellular behavior. In addition to
potential of nanoparticles exhibiting phosphotyrosine sub­ inorganic material, Yoo et al103 synthesized PLGA particles
structures on their surface changed from negative to positive with spherical and elliptical disk geometries and investigated
due to cleavage of negative charges in the form of phosphate the effect of particle shape on rate of particle endocytosis
residues.98 In addition, Shan et al66 reported a simple zwitter­ and their intracellular distribution in endothelial cells. This
ions-based nanoparticle delivery platform, the dense and section briefly summarizes and discusses the relationship
hydrophilic coating of zwitterions endowed the nanoparticles between different shapes of nanoparticles and common bio­
with excellent mucus penetrating ability and affinity with logical endpoints (eg, cell uptake, biological distribution).
epithelial cells, which significantly improved (4.5-fold) the
cellular uptake of nanoparticles, compared with PEGlated Effects of Nanoparticle Morphology on
nanoparticles. Wang et al11 developed a bilayer modification Cellular Uptake in vivo and in vitro
on the surface of mesoporous silica nanoparticles (MSNs) Cell membranes mainly permeate small and non-polar mole­
consisting of polyethylenimine-coated carbon dots (PCD) cules. Most nanoparticles are polar molecules that enter cells
for effective transepithelial absorption and PEG polymers via endocytosis rather than cell diffusion.104 Recently, particle
for improved mucus permeability, which enhanced the stabi­ shape, an important physicochemical property of nanoparti­
lity in the physiological environment, altered cell uptake cles, has gained great attention and been demonstrated experi­
mechanisms, and increased distribution in various intestinal mentally and theoretically to exert a great effect on cellular
sections. uptake behaviors.105 Different morphological features of

submit your manuscript | www.dovepress.com


International Journal of Nanomedicine 2020:15 6303
DovePress

Powered by TCPDF (www.tcpdf.org)


Wang et al Dovepress

nanoparticles affect their endocytosis process. Though some chemistry, and particle composition, which play a dominant
studies have investigated the relationship between shape of role.
nanoparticles and uptake pathways, the revealed results have Endocytosis has two stages: adhesion and internali-
always been inconsistent (Table 3). For instance, it is well zation.109 The shape of nanoparticles has a significant impact
documented that rod-shaped nanoparticles have a lower capa­ on the cell adhesion of a wide variety of nanoparticles. Salatin
city of entering the cells in comparison with their spherical et al110 mentioned that the elongated nanoparticles showed
counterparts.106,107 The data of Wang et al107 also showed the higher efficiency in adhering to the cells rather than the sphe­
same results that sphere-shaped nanoparticles could enter rical nanoparticles. They explained that the curve shape of
mesenchymal stem cells at a faster rate in the initial 4 hours spherical particles allowed a limited number of their binding
than rod-shaped nanoparticles. A possible explanation for their sites to interact with target cell receptors while the elongated
results is that nanorods would seriously destroy the cytoske­ nanoparticles had higher surface area, which facilitated the
multivalent interaction of these particles with cell surface.
leton to cause the reorganization of the cytoskeleton, thereby
Similarly, Zhang et al109 mentioned that the prickly nanoparti­
prolonging the rate of cell internalization and resulting in slow
cles had a higher anchoring amount compared with the round
absorption. In contrast, nanospheres have less impact on the
nanoparticles. The results could be understood as originating
cytoskeleton and eventually get into cells faster.107 However,
from the contact surface area difference of the two types of
Gratton et al108 found that rod-shaped nanoparticles had the
nanoparticles with the plasma membrane. The paper of He and
highest internalization rates compared with spheres, cylinders,
Park111 suggested that a smaller diameter allowed the micro­
and cubes. The results of Banerjee et al6 demonstrated that the
sphere to adhere to cells faster and stronger. Therefore, we can
rod and disc-shaped nanoparticles were internalized 2-fold see that the larger surface area is likely to leadd to a stronger
higher than spherical nanoparticles. The possible reasons for adhesion to the cell. Since strong adhesion between the nano­
the uptake behaviors are governed by cell types, size, surface particles and the cells are required for cells to internalize the
nanoparticles, a larger the surface area implies a higher chance
internalized by cells.112
Table 3 The Different Internalization Pathways Corresponding
NDDS can be capable to enter live cells, often through
to Different Cells and the Main Shapes Associated with Them
several endocytic pathways, namely phagocytosis, macropi­
Cellular Cell Type Nanoparticle Ref nocytosis, clathrin-mediated endocytosis, and caveolin-
Mechanism Shape
mediated endocytosis.113 Phagocytosis only occurs in
Phagocytosis Macrophages Large ARs [116] specialized cells such as macrophages, monocytes, dendritic
Monocytes cells, natural killer cells, and neutrophils, which in turn form
intracellular phagosomes.114 Macropinocytosis is a process by
Dendritic cells
which extracellular fluid and its debris are internalized into
Natural killer a nonspecific manner within large, heterogeneous vesicles,
cells named macropinosomes.115 Clathrin-mediated endocytosis is
Neutrophils initiated by the interaction between ligands and specific recep­
tors. Following entry into the cell, a fraction of internalized
Macropinocytosis Endothelial cells [117]
guest matters would be recycled back to the cell exterior,
Epithelial cells
while the remaining fractions are transferred into lysosomes,
Clathrin-mediated Endothelial cells Sphere-shaped [118] resulting in the degradation of the sequestered cargo material
endocytosis particles by the lysosomal enzymes.116 The caveolae-mediated
Caveolin-mediated The capillary Large ARs [119] endocytosis pathway is characteristic by the evolution of
endocytosis endothelium caveolae-derivatives of the subdomains of sphingolipid and
cholesterol-rich cell membrane fractions, which mediates the
Type I epithelial
cells translocation to the Golgi apparatus, to endoplasmic reticulum
or entered into the endosomal pathway.117
Muscle cells
It has been revealed that the pathway of nanoparticles into
Fibroblasts cells can be regulated by shape. For instance, Hao et al105 used
Abbreviation: AR, aspect ratio. sphere-nanoparticles (NS), short-rod nanoparticles (NSR), and

6304 submit your manuscript | www.dovepress.com International Journal of Nanomedicine 2020:15


DovePress

Powered by TCPDF (www.tcpdf.org)


Dovepress Wang et al

long-rod nanoparticles (NLR) to incubate with Hela cells. shaped particles, stretched from 200 nm sphere-shaped
Their results preliminarily demonstrated that NS particles nanoparticles, were more readily absorbed and transported
preferred to be internalized into cells via the clathrin- by epithelial cells than spheres. Also, Huang et al19 fabri­
mediated pathway, whereas the uptake is shifted to the caveo­ cated and functionalized different shaped MSNs (spheres,
lae-mediated pathway for the particles with large aspect ratios short rods, and long rods) with fluorescein isothiocyanate
(ARs). In addition, Agarwal et al118 mentioned that, although (FITC) and rhodamine B isothiocyanate (RITC) for ima­
macropinocytosis was used by both epithelial and endothelial ging and quantification of MSNs uptake. Then the ability of
cells, epithelial cells uniquely internalized these nanoparticles different shaped MSNs to be transported into model A375
using the caveolae-mediated pathway. Human umbilical vein cells was compared. They found that the number of intra­
endothelial cells, on the other hand, used clathrin-mediated cellular particles varied with particle shape and exhibited
uptake for all shapes and show a significantly higher uptake a strong dependence. Compared with spherical and short
efficiency compared with epithelial cells. Although less sig­ rod-shaped nanoparticles, long rod-shaped nanoparticles
nificantly, it further confirmed that the effect of particle geo­ were easier to internalize. The authors suggested that one
metry is cell type-specific. This shall be one of the reasons for possible explanation was the curvature of different shapes.
the inconsistency between different cells and dependence on The area of contact between rod nanoparticles and the cell
shape. Agarwal et al118 also proposed that when nanoparticle membrane was larger than that of spherical nanoparticles
surface properties and composition were kept constant, each
because the longitudinal axis of rod nanoparticles interacted
cell type could “sense” the nanoscale geometry (both shape
with the cell membrane.
and size) and trigger unique uptake pathways and thus have
Although the main internalization pathways of
different shape-dependent internalization efficiencies.
nanoparticles change with different cells, it could be
Therefore, it is paramount to consider the main intercellular
speculated from the above studies that the curvature of
pathways of different cells in exploring the effects of nano­
nanoparticles and the area of contact with cells play
particle morphology on cellular uptake.
important roles.
Sharma et al119 stretched three different volumes of
Another study said that the morphology dependence of
spherical PS particle, ranging from 0.5–3.6 µm in diameter
cell uptake varied with the size of nanoparticles.101 They
to form prolate or oblate ellipsoids which utilized the film-
believed that larger particles had a general pattern of
stretching method and studied their attachment and inter­
morphological dependence, while smaller nanoparticles
nalization in RAW 264.7 murine macrophages. Regardless
did not have morphological dependence in cellular uptake
of the volume, the order of particle adhesion to the mem­
because small particles tended to change the surface prop­
brane surface was the oblong>oblate>sphere. However, the
erties of nanoparticles. In addition to nanoparticle size, the
order of internalization was the oblate>sphere>oblong.
nanoparticle materials mentioned above also had
Prior to this, Champion and Mitragotri120 also explored
the effect of nanoparticle morphology on macrophage a significant impact on the shape dependence of cell
uptake in a similar approach. They found that regions of uptake. Using Jet and Flash Imprint Lithography (J-FIL)
higher curvature were associated with a larger degree of for top down fabrication of monodisperse, Agarwal et al118
wrapping or phagocytosis. As we all know, nanoparticles fabricated PEG-based discoidal and cuboidal rod-shaped
enter in the system circulation after oral administration nanoparticles. Particles were administered to cells in cul­
where nanoparticles could be absorbed by immune cells in ture at equal total fluorescence intensity. They found that
the blood and various phagocytes in tissues. To avoid being between discoidal and rod-shaped nanoparticles of similar
devoured by various immune cells or macrophages before volume, nanodiscs were more efficiently internalized at
reaching the target tissue, this effect could be minimized by any time points. Nevertheless, Barua et al121 reported
controlling the local shape of the nanoparticles, thereby that for nonspecific hydrophobic PS particles, nanorods
increasing the bioavailability.103 In addition to macro­ and nanodiscs possessed similar uptake in epithelial breast
phages, particle geometry has also been shown to have cancer cells. These differences could highlight the effect of
a significant impact on the context of epithelial drug deliv­ nanoparticle size and modification as well as cell types
ery. Banerjee et al6 investigated the uptake and transport of evaluated and emphasize the importance of material and
smaller stretch nanoparticles in a triple-cell co-culture size composition in understanding nanoparticle-mediated
model of the intestine. They found that rod-shaped and disc- intracellular delivery.

submit your manuscript | www.dovepress.com


International Journal of Nanomedicine 2020:15 6305
DovePress

Powered by TCPDF (www.tcpdf.org)


Wang et al Dovepress

Effects of Nanoparticle Morphology on T02, 2019LZXNYDZ07], the Science and Technology Fund
of Luzhou City (No. 2019-SYF-35), and Science and
GI Transit
Technology Innovation Team from Jiucheng Science and
Geometry not only affects cellular uptake, but also influences
transport across the physiological barriers. Nanorods and Technology Talent Cultivation Plan in Luzhou City (2019-1).
nanospheres were labeled using fluorescence resonance
energy transfer (FRET) molecules to track the in vivo fate Disclosure
of intact nanoparticles accurately.122 Li et al122 found that Yuanyuan Wang reports grants from the Science and
nanorods possessed significantly longer retention time in the Technology Fund for Distinguished Young Scholars of
GI tract compared with nanospheres. Furthermore, nanorods Sichuan Province and Basic Research, the Joint Fund of
exhibited a stronger ability of penetration into space of villi Luzhou City and Southwest Medical University, the
than nanospheres, which was the main reason of longer Science and Technology Fund of Luzhou City, Science and
retention time. In addition, mesenteric lymph transported Technology Innovation Team from Jiucheng Science and
1.75% nanorods within 10 hours, which was more than Technology Talent Cultivation Plan in Luzhou City, the
nanospheres (0.98%). It indicated the nanoparticles shape Scientific Research Foundation of the Education
could affect the movement route or rate. Yu et al123 found Department of Sichuan Province, and a research grant from
that mesoporous silica rods (80×240 nm) as well as calcium Traditional Chinese Medicine Administration in Sichuan
phosphate rods have also been shown to penetrate deeper into Province, during the conduct of the study. The authors report
the mucosal tissue of the GI tract ex vivo compared with no other potential conflicts of interest in this work.
spheres (80 and 140 nm). They guessed that this effect
originated from their rotational diffusion combined with
shear flows and the mesh-like network of mucus enabling
References
deep penetration of the viscoelastic layer. Combined, these 1. Munzone E, Colleoni M. Clinical overview of metronomic che­
motherapy in breast cancer. Nat Rev Clin Oncol. 2015;12
two studies suggested anisotropic and rod-like particles were (11):631–644. doi:10.1038/nrclinonc.2015.131
far superior to targeting the epithelium of the GI tract than 2. Le Z, Chen Y, Han H, et al. Hydrogen-bonded tannic acid-based
anticancer nanoparticle for enhancement of oral chemotherapy.
their spherical equivalents.
ACS Appl Mater Interfaces. 2018;10(49):42186–42197. doi:10.
1021/acsami.8b18979
3. Sarparanta MP, Bimbo LM, Makila EM, et al. The mucoadhesive
Conclusion and Prospective and gastroretentive properties of hydrophobin-coated porous sili­
The physicochemical properties of nanoparticles can signif­ con nanoparticle oral drug delivery systems. Biomaterials.
2012;33(11):3353–3362. doi:10.1016/j.biomaterials.2012.01.029
icantly affect oral bioavailability of drugs. However, oral
4. Ahmad J, Amin S, Rahman M, et al. Solid matrix based lipidic
delivery of drugs is a complex process. And variation in one nanoparticles in oral cancer chemotherapy: applications and
area alone of nanoparticles cannot fully improve the oral pharmacokinetics. Curr Drug Metab. 2015;16(8):633–644.
doi:10.2174/1389200216666150812122128
bioavailability of drugs. Therefore, it is important to under­ 5. van Herwaarden AE, van Waterschoot RA, Schinkel AH. How impor­
stand the role of nanoparticles in the oral delivery. Finally, tant is intestinal cytochrome P450 3A metabolism? Trends Pharmacol
Sci. 2009;30(5):223–227. doi:10.1016/j.tips.2009.02.003
efforts should focus not only on the laboratory synthesis/
6. Banerjee A, Qi J, Gogoi R, Wong J, Mitragotri S. Role of
functionalization, but also on the selection of appropriate nanoparticle size, shape and surface chemistry in oral drug
materials for the preparation of nanoparticles according to delivery. J Control Release. 2016;238:176–185. doi:10.1016/j.
jconrel.2016.07.051
clinical needs, so as to increase the oral absorption and 7. Etrych T, Strohalm J, Chytil P, et al. Biodegradable star HPMA
improve the oral bioavailability of drugs. Here, we have polymer conjugates of doxorubicin for passive tumor targeting. Eur
J Pharm Sci. 2011;42(5):527–539. doi:10.1016/j.ejps.2011.03.001
preliminarily covered the different roles played by different
8. Chen B, Wang X, Zhang Y, et al. Improved solubility, dissolution
properties of nanoparticles in oral drug delivery to provide rate, and oral bioavailability of main biflavonoids from Selaginella
corresponding assistance to researchers in this field. doederleinii extract by amorphous solid dispersion. Drug Deliv.
2020;27(1):309–322. doi:10.1080/10717544.2020.1716876
9. Gong X, Zheng Y, He G, Chen K, Zeng X, Chen Z.
Multifunctional nanoplatform based on star-shaped copolymer
Acknowledgments for liver cancer targeting therapy. Drug Deliv. 2019;26
This study was supported by the basic research fund of the (1):595–603. doi:10.1080/10717544.2019.1625467
Science and Technology Department of Sichuan Province 10. MacEwan SR, Chilkoti A. From composition to cure: a systems
engineering approach to anticancer drug carriers. Angew Chem
(No. 2020YJ0336, 2020YJ0373), the Joint Fund of Luzhou Int Ed Engl. 2017;56(24):6712–6733. doi:10.1002/anie.201
City and Southwest Medical University [No.2017LZXNYD- 610819

6306 submit your manuscript | www.dovepress.com International Journal of Nanomedicine 2020:15


DovePress

Powered by TCPDF (www.tcpdf.org)


Dovepress Wang et al

11. Wang Y, Cui Y, Zhao Y, et al. Effects of surface modification and size 28. Florek J, Caillard R, Kleitz F. Evaluation of mesoporous silica
on oral drug delivery of mesoporous silica formulation. J Colloid nanoparticles for oral drug delivery - current status and perspec­
Interface Sci. 2018;513:736–747. doi:10.1016/j.jcis.2017.11.065 tive of MSNs drug carriers. Nanoscale. 2017;9(40):15252–15277.
12. Mazzaferro S, Bouchemal K, Ponchel G. Oral delivery of anticancer doi:10.1039/C7NR05762H
drugs III: formulation using drug delivery systems. Drug Discov 29. Agoram B, Woltosz WS, Bolger MB. Predicting the impact of
Today. 2013;18(1–2):99–104. doi:10.1016/j.drudis.2012.08.007 physiological and biochemical processes on oral drug
13. Shaikh J, Ankola DD, Beniwal V, Singh D, Kumar MNVR. bioavailability. Adv Drug Deliv Rev. 2001;50(Suppl 1):S41–S67.
Nanoparticle encapsulation improves oral bioavailability of cur­ doi:10.1016/S0169-409X(01)00179-X
cumin by at least 9-fold when compared to curcumin adminis­ 30. Yun Y, Cho YW, Park K. Nanoparticles for oral delivery: targeted
tered with piperine as absorption enhancer. Eur J Pharm Sci. nanoparticles with peptidic ligands for oral protein delivery. Adv
2009;37(3–4):223–230. doi:10.1016/j.ejps.2009.02.019 Drug Deliv Rev. 2013;65(6):822–832. doi:10.1016/j.addr.
14. Swarnakar NK, Jain AK, Singh RP, Godugu C, Das M, Jain S. 2012.10.007
Oral bioavailability, therapeutic efficacy and reactive oxygen 31. Ye W, Chen R, Chen X, et al. AhR regulates the expression of
species scavenging properties of coenzyme Q10-loaded poly­ human cytochrome P450 1A1 (CYP1A1) by recruiting Sp1.
meric nanoparticles. Biomaterials. 2011;32(28):6860–6874. FEBS J. 2019;286(21):4215–4231. doi:10.1111/febs.14956
doi:10.1016/j.biomaterials.2011.05.079 32. Crater JS, Carrier RL. Barrier properties of gastrointestinal mucus
15. Zhao Y, Chen G, Meng Z, et al. A novel nanoparticle drug to nanoparticle transport. Macromol Biosci. 2010;10
delivery system based on PEGylated hemoglobin for cancer (12):1473–1483. doi:10.1002/mabi.201000137
therapy. Drug Deliv. 2019;26(1):717–723. doi:10.1080/10717544. 33. Sigurdsson HH, Kirch J, Lehr CM. Mucus as a barrier to lipo­
2019.1639846 philic drugs. Int J Pharm. 2013;453(1):56–64. doi:10.1016/j.
16. Alam S, Khan ZI, Mustafa G, et al. Development and evaluation of ijpharm.2013.05.040
thymoquinone-encapsulated chitosan nanoparticles for nose-to-brain 34. Varum FJ, Veiga F, Sousa JS, Basit AW. Mucus thickness in
targeting: a pharmacoscintigraphic study. Int J Nanomedicine. the gastrointestinal tract of laboratory animals. J Pharm
2012;7:5705–5718. doi:10.2147/IJN.S35329 Pharmacol. 2012;64(2):218–227. doi:10.1111/j.2042-7158.
17. Ansari MJ, Anwer MK, Jamil S, et al. Enhanced oral bioavail­ 2011.01399.x
ability of insulin-loaded solid lipid nanoparticles: pharmacoki­ 35. Juge N. Microbial adhesins to gastrointestinal mucus. Trends
netic bioavailability of insulin-loaded solid lipid nanoparticles in Microbiol. 2012;20(1):30–39. doi:10.1016/j.tim.2011.10.001
diabetic rats. Drug Deliv. 2016;23(6):1972–1979. doi:10.3109/ 36. Netsomboon K, Bernkop-Schnurch A. Mucoadhesive vs. muco­
10717544.2015.1039666 penetrating particulate drug delivery. Eur J Pharm Biopharm.
18. Ganea GM, Fakayode SO, Losso JN, van Nostrum CF, 2016;98:76–89. doi:10.1016/j.ejpb.2015.11.003
Sabliov CM, Warner IM. Delivery of phytochemical thymoqui­ 37. Strugala V, Allen A, Dettmar PW, Pearson JP. Colonic mucin:
none using molecular micelle modified poly(D, L lactide-co- methods of measuring mucus thickness. Proc Nutr Soc. 2003;62
glycolide) (PLGA) nanoparticles. Nanotechnology. 2010;21 (1):237–243. doi:10.1079/PNS2002205
(28):285104. doi:10.1088/0957-4484/21/28/285104 38. Smith DJ, Gaffney EA, Blake JR. Modelling mucociliary
19. Huang X, Teng X, Chen D, Tang F, He J. The effect of the shape clearance. Respir Physiol Neurobiol. 2008;163(1–3):178–188.
of mesoporous silica nanoparticles on cellular uptake and cell doi:10.1016/j.resp.2008.03.006
function. Biomaterials. 2010;31(3):438–448. doi:10.1016/j. 39. Evans CM, Koo JS. Airway mucus: the good, the bad, the sticky.
biomaterials.2009.09.060 Pharmacol Ther. 2009;121(3):332–348. doi:10.1016/j.
20. Torchilin V. Tumor delivery of macromolecular drugs based on pharmthera.2008.11.001
the EPR effect. Adv Drug Deliv Rev. 2011;63(3):131–135. doi:10. 40. Boegh M, García-Díaz M, Müllertz A, Nielsen HM. Steric and
1016/j.addr.2010.03.011 interactive barrier properties of intestinal mucus elucidated by
21. Nakamura H, Fang J, Maeda H. Development of next-generation particle diffusion and peptide permeation. Eur J Pharm
macromolecular drugs based on the EPR effect: challenges and Biopharm. 2015;95:136–143. doi:10.1016/j.ejpb.2015.01.014
pitfalls. Expert Opin Drug Deliv. 2015;12(1):53–64. doi:10.1517/ 41. Lock JY, Carlson TL, Carrier RL. Mucus models to evaluate the
17425247.2014.955011 diffusion of drugs and particles. Adv Drug Deliv Rev.
22. Fowler R, Vllasaliu D, Trillo FF, et al. Nanoparticle transport in 2018;124:34–49. doi:10.1016/j.addr.2017.11.001
epithelial cells: pathway switching through bioconjugation. Small. 42. Lai SK, Wang YY, Hanes J. Mucus-penetrating nanoparticles for
2013;9(19):3282–3294. doi:10.1002/smll.201202623 drug and gene delivery to mucosal tissues. Adv Drug Deliv Rev.
23. Champion JA, Mitragotri S. Shape induced inhibition of phago­ 2009;61(2):158–171. doi:10.1016/j.addr.2008.11.002
cytosis of polymer particles. Pharm Res. 2009;26(1):244–249. 43. Svensson O, Arnebrant T. Mucin layers and
doi:10.1007/s11095-008-9626-z multilayers-physicochemical properties and applications. Curr
24. Caldorera-Moore M, Guimard N, Shi L, Roy K. Designer nano­ Opin Colloid Interface Sci. 2010;15(6):395–405. doi:10.1016/j.
particles: incorporating size, shape and triggered release into cocis.2010.05.015
nanoscale drug carriers. Expert Opin Drug Deliv. 2010;7 44. Ensign LM, Cone R, Hanes J. Oral drug delivery with polymeric
(4):479–495. doi:10.1517/17425240903579971 nanoparticles: the gastrointestinal mucus barriers. Adv Drug Deliv
25. Li Y, Yue T, Yang K, Zhang X. Molecular modeling of the Rev. 2012;64(6):557–570. doi:10.1016/j.addr.2011.12.009
relationship between nanoparticle shape anisotropy and endocy­ 45. Nordgard CT, Draget KI. Oligosaccharides as modulators of
tosis kinetics. Biomaterials. 2012;33(19):4965–4973. doi:10. rheology in complex mucous systems. Biomacromolecules.
1016/j.biomaterials.2012.03.044 2011;12(8):3084–3090. doi:10.1021/bm200727c
26. Cong VT, Gaus K, Tilley RD, Gooding JJ. Rod-shaped mesopor­ 46. Lieleg O, Ribbeck K. Biological hydrogels as selective diffusion
ous silica nanoparticles for nanomedicine: recent progress and barriers. Trends Cell Biol. 2011;21(9):543–551. doi:10.1016/j.
perspectives. Expert Opin Drug Deliv. 2018;15(9):881–892. tcb.2011.06.002
doi:10.1080/17425247.2018.1517748 47. Chen MC, Sonaje K, Chen KJ, Sung HW. A review of the
27. He B, Lin P, Jia Z, et al. The transport mechanisms of polymer prospects for polymeric nanoparticle platforms in oral insulin
nanoparticles in Caco-2 epithelial cells. Biomaterials. 2013;34 delivery. Biomaterials. 2011;32(36):9826–9838. doi:10.1016/j.
(25):6082–6098. doi:10.1016/j.biomaterials.2013.04.053 biomaterials.2011.08.087

submit your manuscript | www.dovepress.com


International Journal of Nanomedicine 2020:15 6307
DovePress

Powered by TCPDF (www.tcpdf.org)


Wang et al Dovepress

48. Renukuntla J, Vadlapudi AD, Patel A, Boddu SH, Mitra AK. 66. Shan W, Zhu X, Tao W, et al. Enhanced oral delivery of protein
Approaches for enhancing oral bioavailability of peptides and drugs using zwitterion-functionalized nanoparticles to overcome
proteins. Int J Pharm. 2013;447(1–2):75–93. doi:10.1016/j. both the diffusion and absorption barriers. ACS Appl Mater
ijpharm.2013.02.030 Interfaces. 2016;8(38):25444–25453. doi:10.1021/acsami.6b08
49. Bruno BJ, Miller GD, Lim CS. Basics and recent advances in 183
peptide and protein drug delivery. Ther Deliv. 2013;4 67. Ensign LM, Schneider C, Suk JS, Cone R, Hanes J. Mucus
(11):1443–1467. doi:10.4155/tde.13.104 penetrating nanoparticles: biophysical tool and method of drug
50. Luo YY, Xiong XY, Tian Y, Li ZL, Gong YC, Li YP. A review of and gene delivery. Adv Mater. 2012;24(28):3887–3894.
biodegradable polymeric systems for oral insulin delivery. Drug doi:10.1002/adma.201201800
Deliv. 2016;23(6):1882–1891. doi:10.3109/10717544.2015.1052863 68. Ahmad N, Ahmad R, Alam MA, Ahmad FJ. Enhancement of oral
51. Bakhru SH, Furtado S, Morello AP, Mathiowitz E. Oral delivery bioavailability of doxorubicin through surface modified biode­
of proteins by biodegradable nanoparticles. Adv Drug Deliv Rev. gradable polymeric nanoparticles. Chem Cent J. 2018;12(1):65.
2013;65(6):811–821. doi:10.1016/j.addr.2013.04.006 doi:10.1186/s13065-018-0434-1
52. He C, Yin L, Tang C, Yin C. Size-dependent absorption mechan­ 69. Feeney OM, Williams HD, Pouton CW, Porter CJ. ‘Stealth’ lipid-
ism of polymeric nanoparticles for oral delivery of protein drugs. based formulations: poly(ethylene glycol)-mediated digestion
Biomaterials. 2012;33(33):8569–8578. doi:10.1016/j. inhibition improves oral bioavailability of a model poorly water
biomaterials.2012.07.063 soluble drug. J Control Release. 2014;192:219–227. doi:10.1016/
53. Turner JR. Intestinal mucosal barrier function in health and j.jconrel.2014.07.037
disease. Nat Rev Immunol. 2009;9(11):799–809. doi:10.1038/ 70. Wang YY, Lai SK, Suk JS, Pace A, Cone R, Hanes J. Addressing
nri2653 the PEG mucoadhesivity paradox to engineer nanoparticles that
54. Su FY, Lin KJ, Sonaje K, et al. Protease inhibition and absorption “slip” through the human mucus barrier. Angew Chem Int Ed
enhancement by functional nanoparticles for effective oral insulin Engl. 2008;47(50):9726–9729. doi:10.1002/anie.200803526
delivery. Biomaterials. 2012;33(9):2801–2811. doi:10.1016/j. 71. Ejima H, Richardson JJ, Liang K, et al. One-step assembly of
biomaterials.2011.12.038 coordination complexes for versatile film and particle
55. Chuang EY, Lin KJ, Su FY, et al. Calcium depletion-mediated engineering. Science. 2013;341(6142):154–157. doi:10.1126/
protease inhibition and apical-junctional-complex disassembly via science.1237265
an EGTA-conjugated carrier for oral insulin delivery. J Control 72. Shen G, Xing R, Zhang N, Chen C, Ma G, Yan X. Interfacial
Release. 2013;169(3):296–305. doi:10.1016/j.jconrel.2012.11.011 cohesion and assembly of bioadhesive molecules for design of
56. Ling SS, Magosso E, Khan NA, Yuen KH, Barker SA. Enhanced long-term stable hydrophobic nanodrugs toward effective antic­
oral bioavailability and intestinal lymphatic transport of ancer therapy. ACS Nano. 2016;10(6):5720–5729. doi:10.1021/
a hydrophilic drug using liposomes. Drug Dev Ind Pharm. acsnano.5b07276
2006;32(3):335–345. doi:10.1080/03639040500519102 73. Jackson JK, Letchford K. The effective solubilization of hydro­
57. Hanafy A, Spahn-Langguth H, Vergnault G, et al. Pharmacokinetic phobic drugs using epigallocatechin gallate or tannic acid-based
evaluation of oral fenofibrate nanosuspensions and SLN in compar­ formulations. J Pharm Sci. 2016;105(10):3143–3152.
ison to conventional suspensions of micronized drug. Adv Drug doi:10.1016/j.xphs.2016.06.027
Deliv Rev. 2007;59(6):419–426. doi:10.1016/j.addr.2007.04.005 74. Bentley WE, Payne GF. Nature’s other self-assemblers. Science.
58. Zhuang CY, Li N, Wang M, et al. Preparation and characterization 2013;341(6142):136–137. doi:10.1126/science.1241562
of vinpocetine loaded nanostructured lipid carriers (NLC) for 75. Hancock BC, Parks M. What is the true solubility advantage for
improved oral bioavailability. Int J Pharm. 2010;394(1–2):179–­ amorphous pharmaceuticals? Pharm Res. 2000;17(4):397–404.
185. doi:10.1016/j.ijpharm.2010.05.005 doi:10.1023/A:1007516718048
59. Hu L, Tang X, Cui F. Solid lipid nanoparticles (SLNs) to improve 76. Shutava TG, Balkundi SS, Vangala P, et al. Layer-by-layer-coated
oral bioavailability of poorly soluble drugs. J Pharm Pharmacol. gelatin nanoparticles as a vehicle for delivery of natural
2004;56(12):1527–1535. doi:10.1211/0022357044959 polyphenols. ACS Nano. 2009;3(7):1877–1885. doi:10.1021/
60. Das S, Chaudhury A. Recent advances in lipid nanoparticle formu­ nn900451a
lations with solid matrix for oral drug delivery. AAPS PharmSciTech. 77. Xiang S, Yang P, Guo H, et al. Green tea makes polyphenol
2011;12(1):62–76. doi:10.1208/s12249-010-9563-0 nanoparticles with radical-scavenging activities. Macromol
61. Maisel K, Ensign L, Reddy M, Cone R, Hanes J. Effect of surface Rapid Commun. 2017;38(23):1700446. doi:10.1002/marc.2017
chemistry on nanoparticle interaction with gastrointestinal mucus 00446
and distribution in the gastrointestinal tract following oral and 78. Wang X, Chen Y, Dahmani FZ, Yin L, Zhou J, Yao J. Amphiphilic
rectal administration in the mouse. J Control Release. carboxymethyl chitosan-quercetin conjugate with P-gp inhibitory
2015;197:48–57. doi:10.1016/j.jconrel.2014.10.026 properties for oral delivery of paclitaxel. Biomaterials. 2014;35
62. Li H, Chen M, Su Z, Sun M, Ping Q. Size-exclusive effect of (26):7654–7665. doi:10.1016/j.biomaterials.2014.05.053
nanostructured lipid carriers on oral drug delivery. Int J Pharm. 79. Kitagawa S, Nabekura T, Nakamura Y, Takahashi T,
2016;511(1):524–537. doi:10.1016/j.ijpharm.2016.07.049 Kashiwada Y. Inhibition of P-glycoprotein function by tannic
63. Zhao Z, Hu Y, Hoerle R, et al. A nanoparticle-based nicotine acid and pentagalloylglucose. J Pharm Pharmacol. 2007;59
vaccine and the influence of particle size on its immunogenicity (7):965–969. doi:10.1211/jpp.59.7.0008
and efficacy. Nanomedicine. 2017;13(2):443–454. doi:10.1016/j. 80. Ren T, Wang Q, Xu Y, et al. Enhanced oral absorption and
nano.2016.07.015 anticancer efficacy of cabazitaxel by overcoming intestinal
64. de la Fuente M, Csaba N, Garcia-Fuentes M, Alonso MJ. mucus and epithelium barriers using surface polyethylene oxide
Nanoparticles as protein and gene carriers to mucosal surfaces. (PEO) decorated positively charged polymer-lipid hybrid
Nanomedicine (Lond). 2008;3(6):845–857. doi:10.2217/ nanoparticles. J Control Release. 2018;269:423–438. doi:10.10
17435889.3.6.845 16/j.jconrel.2017.11.015
65. Ensign LM, Tang BC, Wang YY, et al. Mucus-penetrating nano­ 81. Li X, Chen D, Le C, et al. Novel mucus-penetrating liposomes as
particles for vaginal drug delivery protect against herpes simplex a potential oral drug delivery system: preparation, in vitro char­
virus. Sci Transl Med. 2012;4(138):138ra179. doi:10.1126/ acterization, and enhanced cellular uptake. Int J Nanomedicine.
scitranslmed.3003453 2011;6:3151–3162. doi:10.2147/IJN.S25741

6308 submit your manuscript | www.dovepress.com International Journal of Nanomedicine 2020:15


DovePress

Powered by TCPDF (www.tcpdf.org)


Dovepress Wang et al

82. Schulz JD, Gauthier MA, Leroux JC. Improving oral drug bioa­ 98. Perera G, Zipser M, Bonengel S, Salvenmoser W, Bernkop-
vailability with polycations? Eur J Pharm Biopharm. 2015;97(Pt Schnurch A. Development of phosphorylated nanoparticles as
B):427–437. doi:10.1016/j.ejpb.2015.04.025 zeta potential inverting systems. Eur J Pharm Biopharm.
83. Bourganis V, Karamanidou T, Samaridou E, Karidi K, 2015;97(Pt A):250–256. doi:10.1016/j.ejpb.2015.01.017
Kammona O, Kiparissides C. On the synthesis of mucus permeat­ 99. Wanger G, Onstott TC, Southam G. Stars of the terrestrial deep
ing nanocarriers. Eur J Pharm Biopharm. 2015;97(Pt subsurface: a novel ‘star-shaped’ bacterial morphotype from
A):239–249. doi:10.1016/j.ejpb.2015.01.021 a South African platinum mine. Geobiology. 2008;6(3):325–330.
84. Pereira de Sousa I, Steiner C, Schmutzler M, et al. Mucus per­ doi:10.1111/j.1472-4669.2008.00163.x
meating carriers: formulation and characterization of highly den­ 100. Young KD. Bacterial morphology: why have different shapes?
sely charged nanoparticles. Eur J Pharm Biopharm. 2015;97(Pt Curr Opin Microbiol. 2007;10(6):596–600. doi:10.1016/j.
A):273–279. doi:10.1016/j.ejpb.2014.12.024 mib.2007.09.009
85. Cánepa C, Imperiale JC, Berini CA, Lewicki M, Sosnik A, 101. Kinnear C, Moore TL, Rodriguez-Lorenzo L, Rothen-
Biglione MM. Development of a drug delivery system based on Rutishauser B, Petri-Fink A. Form follows function: nanoparticle
chitosan nanoparticles for oral administration of interferon-α. shape and its implications for nanomedicine. Chem Rev. 2017;117
Biomacromolecules. 2017;18(10):3302–3309. doi:10.1021/acs. (17):11476–11521. doi:10.1021/acs.chemrev.7b00194
biomac.7b00959 102. Wang C, Ito Y, Pradeep B, Valiyaveettil S. Shape sensitivity on
86. Dyawanapelly S, Koli U, Dharamdasani V, Jain R, Dandekar P. toxicity of gold nanoplates in breast cancer cells. J Nanosci
Improved mucoadhesion and cell uptake of chitosan and chitosan Nanotechnol. 2015;15(12):9520–9530. doi:10.1166/jnn.2015.
oligosaccharide surface-modified polymer nanoparticles for 10780
mucosal delivery of proteins. Drug Deliv Transl Res. 2016;6 103. Yoo JW, Doshi N, Mitragotri S. Endocytosis and intracellular
(4):365–379. doi:10.1007/s13346-016-0295-x distribution of PLGA particles in endothelial cells: effect of
87. Wang M, Zhang Y, Feng J, et al. Preparation, characterization, Particle Geometry. Macromol Rapid Commun. 2010;31
and in vitro and in vivo investigation of chitosan-coated poly (d, (2):142–148. doi:10.1002/marc.200900592
l-lactide-co-glycolide) nanoparticles for intestinal delivery of 104. Foroozandeh P, Aziz AA. Insight into cellular uptake and intra­
exendin-4. Int J Nanomedicine. 2013;8:1141–1154. doi:10.2147/ cellular trafficking of nanoparticles. Nanoscale Res Lett. 2018;13
IJN.S41457 (1):339. doi:10.1186/s11671-018-2728-6
88. Ahmad N, Alam MA, Ahmad R, Umar S, Jalees Ahmad F. 105. Hao N, Li L, Zhang Q, et al. The shape effect of PEGylated
Improvement of oral efficacy of Irinotecan through biodegradable mesoporous silica nanoparticles on cellular uptake pathway in
polymeric nanoparticles through in vitro and in vivo Hela cells. Micropor Mesopor Mat. 2012;162:14–23. doi:10.
investigations. J Microencapsul. 2018;35(4):327–343. doi:10.108 1016/j.micromeso.2012.05.040
0/02652048.2018.1485755 106. Chithrani BD, Ghazani AA, Chan WC. Determining the size and
89. Li H, Lu S, Luo M, Li X, Liu S, Zhang T. A matrix dispersion shape dependence of gold nanoparticle uptake into mammalian
based on phospholipid complex system: preparation, lymphatic cells. Nano Lett. 2006;6(4):662–668. doi:10.1021/nl052396o
transport, and pharmacokinetics. Drug Dev Ind Pharm. 2020;46 107. Wang J, Yang G, Wang Y, et al. Chimeric protein template-induced
(4):557–565. doi:10.1080/03639045.2020.1735408 shape control of bone mineral nanoparticles and its impact on
90. Sanjula B, Shah FM, Javed A, Alka A. Effect of poloxamer 188 mesenchymal stem cell fate. Biomacromolecules. 2015;16
on lymphatic uptake of carvedilol-loaded solid lipid nanoparticles (7):1987–1996. doi:10.1021/acs.biomac.5b00419
for bioavailability enhancement. J Drug Target. 2009;17 108. Gratton SE, Ropp PA, Pohlhaus PD, et al. The effect of particle
(3):249–256. doi:10.1080/10611860902718672 design on cellular internalization pathways. Proc Natl Acad Sci
91. Chaudhary S, Garg T, Murthy RS, Rath G, Goyal AK. Recent U S A. 2008;105(33):11613–11618. doi:10.1073/pnas.0801763
approaches of lipid-based delivery system for lymphatic targeting 105
via oral route. J Drug Target. 2014;22(10):871–882. doi:10.3109/ 109. Zhang B, Feng X, Yin H, et al. Anchored but not internalized:
1061186X.2014.950664 shape dependent endocytosis of nanodiamond. Sci Rep.
92. Trevaskis NL, Charman WN, Porter CJ. Lipid-based delivery 2017;7:46462. doi:10.1038/srep46462
systems and intestinal lymphatic drug transport: a mechanistic 110. Salatin S, Maleki Dizaj S, Yari Khosroushahi A. Effect of the
update. Adv Drug Deliv Rev. 2008;60(6):702–716. doi:10.1016/j. surface modification, size, and shape on cellular uptake of
addr.2007.09.007 nanoparticles. Cell Biol Int. 2015;39(8):881–890. doi:10.1002/
93. Porter CJ, Charman WN. Intestinal lymphatic drug transport: an cbin.10459
update. Adv Drug Deliv Rev. 2001;50(1–2):61–80. doi:10.1016/ 111. He Y, Park K. Effects of the microparticle shape on cellular
S0169-409X(01)00151-X uptake. Mol Pharm. 2016;13(7):2164–2171. doi:10.1021/acs.
94. Khoo SM, Shackleford DM, Porter CJH, Edwards GA, molpharmaceut.5b00992
Charman WN. Intestinal lymphatic transport of halofantrine 112. He Q, Zhang Z, Gao Y, Shi J, Li Y. Intracellular localization and
occurs after oral administration of a unit–dose lipid-based formu­ cytotoxicity of spherical mesoporous silica nano- and
lation to fasted dogs. Pharm Res. 2003;20(9):1460–1465. microparticles. Small. 2009;5(23):2722–2729. doi:10.1002/smll.
doi:10.1023/A:1025718513246 200900923
95. Roger E, Kalscheuer S, Kirtane A, et al. Folic acid functionalized 113. Salatin S, Yari Khosroushahi A. Overviews on the cellular uptake
nanoparticles for enhanced oral drug delivery. Mol Pharm. 2012;9 mechanism of polysaccharide colloidal nanoparticles. J Cell Mol
(7):2103–2110. doi:10.1021/mp2005388 Med. 2017;21(9):1668–1686. doi:10.1111/jcmm.13110
96. Bansil R, Turner BS. Mucin structure, aggregation, physiological 114. Kuhn DA, Vanhecke D, Michen B, et al. Different endocytotic
functions and biomedical applications. Curr Opin Colloid uptake mechanisms for nanoparticles in epithelial cells and
Interface Sci. 2006;11(2–3):164–170. doi:10.1016/j.cocis.2005. macrophages. Beilstein J Nanotechnol. 2014;5:1625–1636.
11.001 doi:10.3762/bjnano.5.174
97. Loretz B, Thaler M, Bernkop-Schnürch A. Role of sulfhydryl 115. Commisso C, Davidson SM, Soydaner-Azeloglu RG, et al.
groups in transfection? A case study with chitosan-NAC Macropinocytosis of protein is an amino acid supply route in
nanoparticles. Bioconjug Chem. 2007;18(4):1028–1035. doi:10. Ras-transformed cells. Nature. 2013;497(7451):633–637. doi:10.
1021/bc0603079 1038/nature12138

submit your manuscript | www.dovepress.com


International Journal of Nanomedicine 2020:15 6309
DovePress

Powered by TCPDF (www.tcpdf.org)


Wang et al Dovepress

116. Watson P, Jones AT, Stephens DJ. Intracellular trafficking pathways 120. Champion JA, Mitragotri S. Role of target geometry in
and drug delivery: fluorescence imaging of living and fixed cells. Adv phagocytosis. Proc Natl Acad Sci U S A. 2006;103
Drug Deliv Rev. 2005;57(1):43–61. doi:10.1016/j.addr.2004.05.003 (13):4930–4934. doi:10.1073/pnas.0600997103
117. Jiang L, Li X, Liu L, Zhang Q. Cellular uptake mechanism and 121. Barua S, Yoo JW, Kolhar P, Wakankar A, Gokarn YR, Mitragotri S.
intracellular fate of hydrophobically modified pullulan nanoparticles. Particle shape enhances specificity of antibody-displaying
Int J Nanomedicine. 2013;8:1825–1834. doi:10.2147/IJN.S44342 nanoparticles. Proc Natl Acad Sci U S A. 2013;110(9):3270–3275.
118. Agarwal R, Singh V, Jurney P, Shi L, Sreenivasan SV, Roy K. doi:10.1073/pnas.1216893110
Mammalian cells preferentially internalize hydrogel nanodiscs 122. Li D, Zhuang J, He H, et al. Influence of particle geometry on
over nanorods and use shape-specific uptake mechanisms. Proc gastrointestinal transit and absorption following oral administration.
Natl Acad Sci U S A. 2013;110(43):17247–17252. doi:10.1073/ ACS Appl Mater Interfaces. 2017;9(49):42492–42502. doi:10.1021/
pnas.1305000110 acsami.7b11821
119. Sharma G, Valenta DT, Altman Y, et al. Polymer particle shape 123. Yu M, Wang J, Yang Y, et al. Rotation-facilitated rapid transport of
independently influences binding and internalization by nanorods in mucosal tissues. Nano Lett. 2016;16(11):7176–7182.
macrophages. J Control Release. 2010;147(3):408–412. doi:10.10 doi:10.1021/acs.nanolett.6b03515
16/j.jconrel.2010.07.116

International Journal of Nanomedicine Dovepress


Publish your work in this journal
The International Journal of Nanomedicine is an international, peer- Journal Citation Reports/Science Edition, EMBase, Scopus and the
reviewed journal focusing on the application of nanotechnology in Elsevier Bibliographic databases. The manuscript management system
diagnostics, therapeutics, and drug delivery systems throughout the is completely online and includes a very quick and fair peer-review
biomedical field. This journal is indexed on PubMed Central, system, which is all easy to use. Visit http://www.dovepress.com/
MedLine, CAS, SciSearch®, Current Contents®/Clinical Medicine, testimonials.php to read real quotes from published authors.
Submit your manuscript here: https://www.dovepress.com/international-journal-of-nanomedicine-journal

6310 submit your manuscript | www.dovepress.com International Journal of Nanomedicine 2020:15


DovePress

Powered by TCPDF (www.tcpdf.org)

You might also like